Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate - MDPI

 
CONTINUE READING
Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate - MDPI
Journal of
           Clinical Medicine

Review
Role of Stromal Paracrine Signals in Proliferative
Diseases of the Aging Human Prostate
Kenichiro Ishii 1,2, *    ID
                               , Sanai Takahashi 3 , Yoshiki Sugimura 2 and Masatoshi Watanabe 1,3     ID

 1    Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
      mawata@doc.medic.mie-u.ac.jp
 2    Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine,
      Tsu, Mie 514-8507, Japan; sugimura@clin.medic.mie-u.ac.jp
 3    Laboratory for Medical Engineering, Division of Materials Science and Chemical Engineering,
      Graduate School of Engineering, Yokohama National University, Yokohama, Kanagawa 240-8501, Japan;
      sanai.takahashi@gmail.com
 *    Correspondence: kenishii@clin.medic.mie-u.ac.jp; Tel.: +81-59-232-1111
                                                                                                      
 Received: 13 March 2018; Accepted: 28 March 2018; Published: 2 April 2018                            

 Abstract: Androgens are essential for the development, differentiation, growth, and function of the
 prostate through epithelial–stromal interactions. However, androgen concentrations in the hypertrophic
 human prostate decrease significantly with age, suggesting an inverse correlation between androgen
 levels and proliferative diseases of the aging prostate. In elderly males, age- and/or androgen-related
 stromal remodeling is spontaneously induced, i.e., increased fibroblast and myofibroblast numbers,
 but decreased smooth muscle cell numbers in the prostatic stroma. These fibroblasts produce not only
 growth factors, cytokines, and extracellular matrix proteins, but also microRNAs as stromal paracrine
 signals that stimulate prostate epithelial cell proliferation. Surgical or chemical castration is the standard
 systemic therapy for patients with advanced prostate cancer. Androgen deprivation therapy induces
 temporary remission, but the majority of patients eventually progress to castration-resistant prostate
 cancer, which is associated with a high mortality rate. Androgen deprivation therapy-induced stromal
 remodeling may be involved in the development and progression of castration-resistant prostate cancer.
 In the tumor microenvironment, activated fibroblasts stimulating prostate cancer cell proliferation are
 called carcinoma-associated fibroblasts. In this review, we summarize the role of stromal paracrine
 signals in proliferative diseases of the aging human prostate and discuss the potential clinical applications
 of carcinoma-associated fibroblast-derived exosomal microRNAs as promising biomarkers.

 Keywords: proliferative diseases of the aging human prostate; epithelial–stromal interactions;
 stromal remodeling; stromal paracrine signals; carcinoma-associated fibroblast-derived
 exosomal microRNAs

1. Introduction
     The prostate is a male accessory sex gland found only in mammals. Its main function is to produce
a major proportion of the seminal fluid. Androgens are essential for the development, differentiation,
growth, and function of the prostate [1].
     The prostate develops from the urogenital sinus and is induced by embryonic mesenchyme,
fibroblasts, and myofibroblasts during its development and differentiation [2]. Ductal morphogenesis,
epithelial differentiation, and proliferation/apoptosis are regulated by androgens acting through the
stromal androgen receptor (AR). As summarized in Figure 1, androgen-mediated stromal paracrine
signals support normal epithelial differentiation and function [3]. As stromal paracrine signals for
epithelial cells, stromal components such as fibroblasts, myofibroblasts, and smooth muscle cells secrete

J. Clin. Med. 2018, 7, 68; doi:10.3390/jcm7040068                                   www.mdpi.com/journal/jcm
Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate - MDPI
J. Clin. Med. 2018, 7, 68                                                                               2 of 21

a number of growth factors, cytokines, extracellular matrix (ECM) proteins, and microRNAs (miRNAs).
Thus, androgen-mediated epithelial–stromal interactions play crucial roles in ductal morphogenesis
and epithelial differentiation in the development of the prostate [4,5].

      Figure 1. Epithelial–stromal interactions in the developing prostate. Bone marrow-derived
      mesenchymal stem cells may differentiate into fibroblasts, myofibroblasts, and smooth muscle cells
      in prostatic stroma. Fibroblasts and myofibroblasts secrete mitogens to stimulate the proliferation
      of undifferentiated cells. Smooth muscle cells secrete morphogens to maintain the functional
      differentiation of adult epithelial cells. AR: androgen receptor; Shh: sonic hedgehog.

     In the adult prostate, stroma composed of smooth muscle cells interact with epithelial cells.
These epithelial–stromal interactions maintain the functional differentiation and growth/quiescence of
epithelial cells [6]. In adults, androgens act on AR-positive epithelial cells to establish their functional
differentiation into luminal secretory epithelial cells [7]. Moreover, epithelial AR is responsible for
maintaining the differentiated phenotype and overall homeostasis of the prostate glands [6].
     Androgens also act on epithelial cells to produce growth factors, cytokines, and miRNAs that
mediate stromal functions, including proliferation and differentiation [8–10]. On the other hand,
androgens act on smooth muscle cells to produce a number of morphogens critical for maintaining
the differentiation and supporting the proliferation of epithelial cells [11]. Furthermore, under the
influence of androgens, stromal cells produce mitogens that stimulate the proliferation of epithelial
cells [12]. Thus, these androgen-mediated paracrine effects between epithelial and stromal cells
maintain homeostasis in the adult prostate [13,14]. An interesting feature of the adult prostate is that
the tissue does not actively proliferate, and there is little cellular turnover even in the presence of
growth-stimulating androgens. Moreover, the incidences of proliferative diseases of the aging human
prostate, including benign prostatic hyperplasia (BPH) and prostate cancer (PCa), apparently increase
with advancing age, despite decreasing androgen levels [15].
     The androgen status affects prostatic structure and components. In the absence of androgens,
prostatic structural regression is mainly attributed to a functional decrease in the secretory activity of
luminal epithelial cells and to a reduction in the number of luminal epithelial cells by apoptosis [16,17].
Structural alterations also occur in the stroma following castration and are characterized by the
replacement of smooth muscle cells by fibroblasts or myofibroblasts, called stromal remodeling [18,19].
Evidence suggests that castration-induced stromal remodeling is accompanied by functional
transformation of the prostatic stromal environment [20]. In luminal epithelial cells, prostatic secretory
functions and numbers are dramatically decreased in the absence of androgen, while basal epithelial
cells remain intact to form a continuous layer lining the atrophied prostate acini in the absence
of androgen [18,19]. Interestingly, this regression process after castration is reversible, because
androgen replacement leads to complete regeneration of the prostate with secretory function [21–24].
Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate - MDPI
J. Clin. Med. 2018, 7, 68                                                                           3 of 21

The mechanisms of prostatic regeneration may include the participation of prostate stem/progenitor
cells with self-renewing ability [25,26]. For example, a single transplanted stem cell is sufficient to
regenerate the prostate in vivo [24].

2. Benign Prostatic Hyperplasia (BPH)
     BPH is a common prostatic disease in elderly males. The incidence of BPH increases with age
and more rapidly than that of PCa [27]. Notably, most PCas arise in the prostate concomitant with
BPH elsewhere [28].
     BPH is frequently observed in middle-aged and elderly males; its prevalence is only 8% during the
fourth decade of life, but more than 70% during the seventh decade [29]. Its major manifestations are
lower urinary tract symptoms. Because it is benign, severe complications are rare. However, from the
standpoint of preserving quality of life, it is essential to alleviate the symptoms of BPH. Regarding the
natural history of BPH, Berry et al. reported that prostate weight increases sharply as androgen
levels increase before adolescence, although the rate of increase becomes much slower after the age of
30 years [29]. However, prostate weight begins to increase again over the age of 60 years, with a sharp
increase in the 70s. In an analysis of Japanese cases reported by Fujikawa et al., prostate weight tended
to increase sharply from birth to the third decade of life and thereafter increased gradually, with no
sharp increase after age 60 [30]. These two reports suggest that prostate weight is lower in Japanese
than in Western individuals, and that prostate weight gain is less in Japanese males.
     With regard to the size of the prostate, Ishigooka et al. reported a correlation between prostate
volume and increased fibrous tissue in a histological examination of surgical specimens obtained by
transurethral resection of the prostate (TURP), suggesting that fibrous tissue plays an important role
in BPH [31]. Ichiyanagi et al. performed a histological examination of TURP specimens and found that
the proportions of stroma and gland cavities in the prostate differed little between patients with and
those without disturbance of the lower urinary tract passage, while the proportion of smooth muscle
in prostatic stroma was significantly lower in patients with a disturbance in the lower urinary tract
passage [32].

3. Alteration of Stromal Structure by α1 -Adrenoceptor Antagonists (α1 -Blockers)
      At present, BPH is treated by drug therapies, surgery (primarily TURP), or less-invasive therapies
using lasers, stents, and other means. α1 -adrenoceptor antagonists (α1 -blockers) are often used as
first-line drugs [33]. Even at present, 20 years since their clinical use was introduced, α1 -blockers
are considered useful in the treatment of BPH. They suppress functional contraction of prostatic
smooth muscle cells by blocking signaling from the α1 -adrenoceptors distributed in this muscle.
α1 -blockers are highly efficacious in alleviating lower urinary tract symptoms and are therefore
frequently used clinically.
      Smith et al. found that the contractility of prostatic smooth muscle cells decreased following the
inhibition of adrenergic receptor signaling by α1 -blockers [34]. Their finding suggests that α1 -blockers
induce dedifferentiation of smooth muscle cells into fibroblasts and myofibroblasts and may thus
induce alteration of tissue architecture. Moreover, Justulin et al. reported that when rats were given
repeated doses of doxazosin, an α1 -blocker, they exhibited an accumulation of collagen fibers in the
prostatic stroma and a reduction in smooth muscle cells [35]. Based on this finding, they suggested the
possibility that continued oral treatment with doxazosin would alter stromal structure and components,
reducing the efficacy of α1 -blockers [35]. These views were endorsed by the findings of our study,
in which treatment with α1 -blockers resulted in alteration of stromal structure and components [36].
However, the mechanisms responsible for such changes have not yet been elucidated. Justulin et al.
suggested that transforming growth factor (TGF)-β is an intracellular transmitter potentially involved
in these stromal alterations [35]. TGF-β is a cytokine with diverse functions. In the ECM, fibroblasts,
osteoblasts, odontoblasts, and other types of cells induce the expression of COL1A1 and COL1A2 (genes
J. Clin. Med. 2018, 7, 68                                                                             4 of 21

encoding type I collagen) in the presence of TGF-β, resulting in the stimulation of type I collagen
formation [37].

4. Aberrant Activation of Epithelial-Stromal Interactions in BPH
     Deregulation of epithelial–stromal interactions is considered to be responsible for the initiation
and/or promotion of proliferative diseases of the aging human prostate, including BPH and PCa [38,39].
As shown in Figure 2, stromal nodules associated with BPH are composed of fibroblasts, myofibroblasts,
and smooth muscle cells [40]. As a potential developmental mechanism of BPH, Pradhan et al. reported
that nodular hyperplasia originates as an early stromal nodule, usually by the side of the urethra,
stimulating the duct in its vicinity to proliferate [41].

      Figure 2. Stromal changes associated with proliferative diseases of the aging human prostate.
      Surgical tissue specimens obtained from patients with BPH or PCa were histologically stained with
      Masson’s trichrome and immunostained with an anti-tenascin-C antibody. Masson’s trichrome stained
      smooth muscle cells pink and fibroblasts/myofibroblasts blue. BPH: benign prostatic hyperplasia;
      PCa: prostate cancer. Magnification 400×.

     According to the hypothesis proposed by McNeal, stromal nodules are the result of the
reappearance of embryonic ductal morphogenesis, and BPH develops through a change in stromal
differentiation into the fetal phenotype [42]. Norman et al. suggested that in mice, the fetal stroma
reacts with post-developmental prostate epithelia, resulting in the formation of new prostate tissue [43].
Their report favors the hypothesis of McNeal and suggests that various growth factors, cytokines,
ECM proteins, and miRNAs involved in aberrant activation between epithelial and stromal cells are
related to the onset of BPH.

5. Effects of Sex Steroid Hormone Status on Basal Epithelial Cell Behavior in the Prostate
     The prostate contains two major epithelial cell types: luminal and basal epithelial cells.
Basal epithelial cells are critical for maintaining ductal integrity and regulating both the survival
and apoptosis of luminal epithelial cells [44–48]. Moreover, prostate progenitor and stem cells have
been identified within the basal compartment [49].
     An increase in basal epithelial cells is referred to as basal cell hyperplasia, which is defined as
basal cell proliferation composed of two or more layers of small cells with scant cytoplasm presenting
as glands or solid nests [50]. Basal cell hyperplasia is occasionally a component of untreated BPH,
which arises in the transition zone of the human prostate [50–52]. Alteration of the proliferation of
basal epithelial cells in the peripheral zone was suggested to be involved in the initiation and early
progression of PCa [53]. Clinically, androgen ablation by antiandrogen therapy for patients with
advanced PCa results in basal cell hyperplasia with variable focal squamous metaplasia localized
diffusely throughout benign prostate tissue [54].
J. Clin. Med. 2018, 7, 68                                                                              5 of 21

      Several studies have reported that androgen ablation leads to the death of luminal secretory
epithelial cells, while basal cells, an AR-negative population, survive [18,19]. The androgen
concentration in the hypertrophic human prostate decreases significantly with age [15]. In the
dorsolateral prostate (DLP) of senescence-accelerated mice deficient in androgen, stromal fibrosis,
the presence of atypical glandular epithelial cells, and cribriform glandular deformities were observed
as age-related alterations [55]. In the canine prostate, the effects of androgen ablation on basal
epithelial cells and luminal epithelial cells are associated with a marked increase in the stromal
fibromuscular compartment, which demonstrates impaired differentiation [18]. The age-related
expansion of proliferating acinar basal epithelial cell populations, mediated by sex steroid hormones,
is a key factor in the pathogenesis of canine prostatic hyperplasia [56]. These findings indicate
that an aberrant proliferation of epithelial cells may be related to the concentration of androgen in
the prostate.
      In our lab, Kato et al. demonstrated that the number of basal epithelial cells in the mouse prostate
was affected by changes in androgen status [19]. Interestingly, the number of basal epithelial cells was
increased in the absence of androgen and returned to baseline levels following androgen replacement.
We propose that these proliferative alterations observed in the regressed prostate may compensate for
the loss of luminal epithelial cells to maintain residual ductal components. Androgen replacement after
castration drives complete regeneration of the gland, which is derived entirely from the proliferation
and differentiation of surviving cells, including basal epithelial cells [57].
      In the castrated mouse prostate, the levels of specific growth factors are increased in the setting of
androgen withdrawal [19,58–60]. To examine how specific growth factors affect prostatic structure,
we analyzed androgen-status-dependent changes in the gene expression of growth factors in involuted
mouse prostates [19]. After castration, the mRNA expression levels of specific growth factors, such as
Fgf2, Fgf7, Hgf, Tgfa, and Tgfb, were relatively abundant in whole mouse DLPs. In embryos, these
growth factors stimulate the proliferation of epithelial cells and play an important role in prostate
development and ductal morphogenesis [61,62]. In organ culture experiments, abnormal basal
epithelial proliferation was recapitulated in the absence of dihydrotestosterone (DHT). The proliferation
of basal epithelial cells in the absence of DHT was suppressed by treatment with a fibroblast
growth factor (FGF) receptor inhibitor (PD173074). Moreover, FGF2 treatment directly stimulated
the proliferation of basal epithelial cells. Finally, we concluded that the FGF2–FGF receptor signaling
cascade in the prostate gland may be one pathway that stimulates the proliferation of basal epithelial
cells in the absence of androgens [19].
      In the prostate, squamous metaplasia, which involves an increase in the number of basal epithelial
cells [63], is induced by estrogens, including 17β-estradiol (E2 ), diethylstilbestrol (a synthetic estrogen),
and bisphenol A (an endocrine-disrupting chemical), which has weak estrogenic activity [64–67].
Estrogen action in the prostate is mediated through estrogen receptors α (ERα) and β (ERβ).
In particular, estrogen-induced squamous metaplasia requires ERα signaling in both epithelial and
stromal cells [63]. In neuroblastoma cells, E2 induced synthesis of transforming growth factor α (TGFα)
via ERα, and TGFα in turn, induced cell proliferation [68]. In another report, long-term treatment
with gonadal steroids, testosterone, and E2 induced dysplasia and enhanced TGFα and epidermal
growth factor (EGF) receptor expression in the normal rat prostate [69]. Regarding the role of TGFα,
we demonstrated that the number of basal epithelial cells in the prostate of TGFα-overexpressing mice
was increased [70]. These reports support that TGFα may be a second messenger in estrogen signaling,
mediating estrogen-induced basal epithelial cell growth in the prostate.

6. Prostate Cancer (PCa)
     The number of males diagnosed with PCa is increasing all over the world [71]. Most patients with
early-stage PCa can be treated by the appropriate therapy such as radical prostatectomy or irradiation.
On the other hand, androgen deprivation therapy (ADT) is the standard systemic therapy given to
patients with advanced PCa. ADT induces temporary remission, but the majority of patients eventually
J. Clin. Med. 2018, 7, 68                                                                           6 of 21

progress to castration-resistant PCa (CRPC), which is associated with a high mortality rate [72,73].
CRPC, a heterogeneous disease, exhibits varying degrees of androgen sensitivity. Once PCa cells
lose their sensitivity to ADT, effective therapies are limited. The progression of PCa cells from local
invasion to distant metastasis and androgen insensitivity may be influenced by alterations in the tumor
microenvironment, AR mutations, overexpression of growth factors and their receptors, and secretion
of ECM proteins and miRNAs, leading to selection of cells with higher aggressive potential [10,74].
Understanding the biological changes that occur in the androgen-insensitive state may help identify
new pathways as drug targets.
     Acute loss of AR function after ADT is associated with not only apoptosis and a reduction in
prostate-specific antigen (PSA) secretion, but also promotion of AR-independent growth in PCa cells.
Disruption of androgen signaling by ADT may result in deregulation of cell cycle control, which
could contribute to carcinogenesis [75]. Nelson et al. described four molecular-state frameworks
for AR activation in PCa after ADT as follows: state 1, endocrine androgen dependent and AR
dependent; state 2, intracrine androgen dependent and AR dependent; state 3, androgen independent
and AR dependent; and state 4, androgen independent and AR independent [76]. State 4 is considered
the fatal stage, at which AR signaling is abolished, and neuroendocrine (NE) differentiation occurs.
Burchardt et al. reported that LNCaP cells inoculated into castrated mice induced a significant
increase in NE cells compared with inoculation into intact mice [77]. Although the mechanism of NE
transdifferentiation induced by androgen deprivation of AR-dependent PCa cells has been investigated,
the mechanism mediating NE differentiation is still not clear [78].

7. Tumor–Stromal Interactions in PCa
     In adult prostate homeostasis, epithelial–stromal interactions maintain functional differentiation
and growth/quiescence [79]. In contrast, the deregulation of epithelial–stromal interactions is
considered to play a critical role in the initiation and/or promotion of carcinogenesis [38,39]. In the
tumor microenvironment, aberrant activation between cancer cells and stromal cells significantly
contributes to the progression of human cancers including prostate, breast, and colon [80–84].
     Solid tumors are highly complex and heterogeneous and are composed of epithelial cancer
cells infiltrating into the surrounding tumor stroma, called reactive stroma and comprising
carcinoma-associated fibroblasts (CAFs) [85,86]. PCa is interesting because of the multi-focal and
heterologous progression of primary tumors [87]. Recent in vivo studies have demonstrated that the
heterogenous stromal compartment of the prostate contains multiple populations of fibroblasts that are
associated with tumorigenesis [88,89]. Clinically, reactive stromal grading in radical prostatectomies or
biopsies is a predictor of recurrence; a high reactive stromal grade is associated with lower biochemical
recurrence-free survival rates compared with a low grade [90,91].
     CAFs surrounding cancer cells secrete a number of growth factors, cytokines, ECM proteins,
and miRNAs to support the survival and proliferation of cancer cells in a paracrine fashion [39,92].
Tsunoda et al. reported that the expression of periostin, a stromal biomarker in PCa, in the CAFs of PCa
tissues was increased during the early stages of the disease (Gleason score 6–7) and was significantly
correlated with the degree of malignancy [93]. Up-regulation of collagen I, tenascin-C, and TGF-β has been
observed in PCa specimens [85]. In addition, the phenotype of cultured prostate CAFs expressing high
levels of CD90, a marker of mesenchymal stem cells (MSCs), was associated with greater tumor promotion
than that of cells with low CD90 expression [94]. CAFs with high CD90 levels expressed higher levels of
many genes associated with tumor promotion, including TGF-β, the angiogenic factors vascular endothelial
growth factor (VEGF) and FGF2, and the cytokines interleukin (IL)-6 and chemokine (C-X-C motif) ligand
12. Several studies have shown that CAFs isolated from PCa specimens are strongly heterogenous and
have specific biochemical characteristics [95–98]. In our lab, we reported that primary cultured PCaSC-8
and/or PCaSC-9 cells derived from different human PCa specimens displayed a significantly higher
mRNA expression of COL1A1, TNC, EGF, FGF2, FGF7, HGF, and IGF1 [96]. The role of ECM proteins
in tumorigenesis includes effects on epithelial polarity and angiogenesis [99]. In contrast, CAF-derived
J. Clin. Med. 2018, 7, 68                                                                            7 of 21

growth factors are predominantly stimulators of cancer cell proliferation and play a part in promoting the
carcinogenic process [100]. Importantly, the expression profiles of CAF-related genes were heterogenous
between PCaSC-8 and PCaSC-9 cells, suggesting that the biochemical characteristics of different human PCa
specimen-derived CAFs are strongly heterogenous. Previous studies showed differences in biochemical
characteristics between human PCa specimen-derived CAFs and adjacent normal fibroblasts [95,98].
      The adult prostate contains an abundance of stromal components, mainly consisting of
well-differentiated smooth muscle cells. In prostate tumors, however, fibroblasts surrounding cancer
cells are associated with not only the initiation of cancer cells, but also tumor growth and progression to
androgen independence [101–103]. Olumi et al. demonstrated stroma-induced malignant transformation,
indicating that fibroblasts surrounding epithelial cells play an important role in PCa development [38].
Other groups reported that co-inoculation of PCa cells with fibroblasts increased tumorigenicity and
potentially angiogenesis promotion [84,101,104]. Therefore, the inhibition of tumor–stromal interactions
may exert a synergistic effect with the suppression of cancer cell growth on tumor control.
      During cancer progression, stromal changes result in a decreased prevalence of smooth
muscle cells (Figure 2). Tumor stroma surrounding cancer cells is enriched in fibroblasts and
myofibroblasts [105]. Several studies have demonstrated the presence of myofibroblastic cells with the
capacity to contribute ECM proteins and collagenous components to the reactive stroma surrounding
tumors [85,106,107]. Myofibroblasts, which share characteristics with fibroblasts and smooth muscle
cells, are activated fibroblasts typically found at sites of pathologic tissue remodeling, such as wound
healing [108,109]. In cancers, myofibroblasts in reactive stroma show irregularities in length and thickness
compared with fibroblasts.

8. PCa Cell Lines with Different Levels of Androgen Sensitivity
      Decrease or loss of androgen sensitivity in PCa is a clinical concern. Many studies on CRPC have used
androgen-insensitive PCa cell lines, such as PC-3 and DU145 cells, which do not express AR. These cell
lines originated from highly anaplastic tumors from different metastatic sites such as the bone and
brain [110,111]. Both the PC-3 and DU145 cell lines are strongly different in terms of aggressiveness
compared with the androgen-sensitive AR-positive LNCaP line, which originated from lymph node
metastasis [112]. Comparisons between the androgen-sensitive LNCaP and androgen-insensitive PC-3
and DU145 cell lines may not be relevant to the acquisition of androgen insensitivity in clinical PCa,
because many clinical androgen-insensitive PCa cases express AR. A more precise model of clinical
cancer requires, at the very least, an androgen-insensitive AR-positive cancer cell line. To compare the
biochemical characteristics of androgen-insensitive and androgen-sensitive PCa cells, we generated three
sublines from androgen-sensitive LNCaP cells: E9 and F10 cell lines (low androgen sensitivity) [113,114]
and androgen-insensitive AIDL cells [115]. The parental LNCaP cells and the derived E9, F10, and AIDL
cells express similar levels of AR protein, but androgen-dependent PSA secretion is only detected in
LNCaP cells [116]. Moreover, we have shown that recombination of E9 cells or AIDL cells with embryonic
rat urogenital sinus mesenchyme (UGM) promoted tumor progression in vivo even under androgen
ablation [116].

9. Origins of Cell Populations Composed of Tumor Stroma
     In various solid tumors, including those of the breast, colon, lung, and prostate, tumor stroma
including CAFs has been implicated in tumor growth, progression, angiogenesis, and metastasis [105,117].
The origin of CAFs has not been well defined, but Mishra et al. reported that bone marrow-derived MSCs
could be a candidate cell origin for CAFs in solid tumors [118]. They showed that upon exposure
to conditioned medium from human breast cancer MDAMB231 cells, MSCs became activated and
exhibited a CAF-like myofibroblastic phenotype. Several reports demonstrated that circulating MSCs in
the bloodstream are recruited and localized to developing tumors [119,120], suggesting that certain factors
secreted from tumors recruit MSCs to solid tumors. Although the specific tumor-derived factors have
not been identified, Wang et al. reported that the differentiation of MSCs into myofibroblasts is regulated
J. Clin. Med. 2018, 7, 68                                                                                  8 of 21

by TGF-β [121]. Verona et al. reported that TGF-β stimulated prostatic stromal cells to express several
genes related to myofibroblastic differentiation, including COL11A1, TNC, and ACTA2, and promoted
reactive stroma formation and carcinoma growth in vivo [101]. These results suggest that MSCs, recruited
by tumors and activated by TGF-β in solid tumors, may be a potential candidate source of CAFs.
       Most human cancers result from an accumulation of somatic mutations arising in epithelial cells.
The behavior of cancer cells is influenced by the tumor microenvironment including CAFs, ECM proteins,
blood vasculature, and inflammatory cells. At present, it is not clear how cancer cells influence the
generation of reactive stroma, or to what extent the reactive stroma is a result of phenotypic changes in
resident cells versus cells recruited from other sites.
       The CAFs in tumor tissues are generated in response to specific paracrine signals from adjacent cancer
cells. Although the origin of CAFs has not been determined, normal fibroblasts are abundant in prostate
tissues. The variety of CAFs present may reflect different cell lineages or site-specific induction [108].
In PCa specimens, different malignant cell types are distributed heterogeneously throughout the tissues.
This pathological feature led us to hypothesize that the generation of CAFs in PCa may be dependent on
the biochemical characteristics of adjacent cancer cells. In our lab, Ishii et al. demonstrated that normal
fibroblasts co-cultured with PCa cells become activated and exhibit biochemical characteristics of CAFs in
a heterogenous manner [96]. Our results suggest that the heterogenous induction of CAF-like differentiation
might be strongly dependent on the biochemical characteristics of adjacent cancer cells (Figure 3). In that
study, we used three cancer cell lines for in vitro co-culture experiments: the androgen-sensitive LNCaP
cell line and its derivative sublines, low-androgen-sensitive E9 cells, and androgen-insensitive AIDL cells.
Co-cultures of commercially available prostate stromal cells and PCa cells in vitro changed the cytogenetic
and biochemical profiles of stromal cells in a cancer cell line-specific manner. Our previous studies
demonstrated that E9 and AIDL cells acquire a more aggressive phenotype in vitro and in vivo compared
with the parental LNCaP cells [113,116,122,123]. However, we have not investigated in detail the differences
in biochemical characteristics among these three cell lines. In our lab, we have shown that the biochemical
characteristics of prostate stromal cells co-cultured with E9 cells, but not LNCaP or AIDL cells, resembled
those of PCaSC-8 and PCaSC-9 cells [96]. This supports that heterogenous induction of a CAF-like
phenotype may be strongly dependent on the specific characteristics of adjacent cancer cells. Identifying the
mechanisms underlying the heterogenous induction of CAF-like differentiation in normal fibroblasts is
an initial step toward designing CAF-targeted therapies for the treatment of PCa. Future studies will
attempt to identify the specific profiles of the factors responsible for the CAF phenotype.

      Figure 3. Induction of carcinoma-associated fibroblasts in the tumor stroma of prostate cancer.
      PCa cells induce CAF-like differentiation in normal fibroblasts. Importantly, heterogenous induction
      of CAF-like differentiation may be strongly dependent on the biochemical characteristics of PCa cells.
      AR: androgen receptor; CAFs: carcinoma-associated fibroblasts; PCa: prostate cancer.
J. Clin. Med. 2018, 7, 68                                                                         9 of 21

10. Characteristics of Cell Populations Composed of Tumor Stroma
      To investigate the effects of stromal components on tumorigenesis, Gleave et al. demonstrated
that organ-specific fibroblasts were responsible for prostate tumor growth in vivo, i.e., LNCaP tumor
formation was most induced by human bone fibroblasts, followed by rat UGM and Noble rat prostatic
fibroblasts, but not by NIH-3T3, normal rat kidney, or human lung CCD16 fibroblasts [124]. In contrast,
UGM has a normalizing effect on Dunning PCa cells [125]. Of interest, cancer cells in tumors have the
ability to induce adjacent normal smooth muscle cells to exhibit a reactive myofibroblastic phenotype
with some features similar to those of UGM [84]. Thus, although the tumor–stromal interactions
are strongly complicated, it is obvious that the tumor and stroma affect each other and cooperate in
tumor progression.
      Reactive stroma or CAFs adjacent to cancer cells secrete growth factors such as EGF family
members, FGFs, insulin-like growth factors (IGFs), and TGF-β, which are involved in development
and cancer progression, invasion, metastasis, and angiogenesis [102,126–129]. These growth factors
are also produced by UGM [116,123,130]. UGM is composed of undifferentiated fibroblasts that induce
instructive and permissive development and differentiation in the prostate. In our study, UGM shows
androgen-dependent cell growth [116]. Interestingly, the expression of FGF2 and IGF1 mRNA was
dramatically decreased in UGM cultured under androgen starvation, while FGF7 mRNA was not
influenced by androgen status. This suggests that FGF7/keratinocyte growth factor (KGF) may act as
an androgen-independent stromal paracrine signal. Planz et al. reported that FGF7/KGF stimulated
the proliferation of LNCaP cells in the presence of the anti-androgenic agent flutamide, showing that
FGF7/KGF-induced cell proliferation in LNCaP cells is independent of cellular AR signaling [131].
Our results support the idea that androgen-independent stromal paracrine signaling by FGF7/KGF
may bypass the functionally inactive AR and promote the proliferation of androgen-insensitive PCa
cells during ADT.
      Interestingly, Halin et al. demonstrated that androgen-insensitive PCa cells respond to castration
when growing in an androgen-dependent prostate environment [132]. When androgen-insensitive
AT-1 PCa cells were injected into the ventral prostates of Copenhagen rats, an androgen-dependent
environment, castration reduced AT-1 tumor growth and vascular density in the tissue surrounding
the tumor. These data demonstrated the importance of the cancer cell microenvironment for the
action of androgens, i.e., the tumor growth of androgen-insensitive PCa cells was regulated by
androgen-mediated stromal paracrine signals including growth factors, cytokines, and miRNAs.
However, the mechanisms by which androgen-insensitive PCa cells respond to stromal paracrine
signals under low-androgen conditions are not yet fully understood.
      The appearance of CAFs and ECM deposition in tumor tissues has been reported in many types
of human cancers [108,109]. In particular, CAFs provide potentially oncogenic signals; CAF-derived
tenascin-C and TGF-β participate in the acceleration of cancer cell invasion, and CAF-derived growth
factors and angiogenic factor VEGF can stimulate cancer progression, including angiogenesis.
      During primary cancer progression, CAFs communicate with cancer cells through the secretion of
growth factors, cytokines, ECM proteins, and miRNAs [133,134]. For example, CAF-derived TGF-β,
EGF, FGFs, VEGF, matrix metalloproteinases, and a number of other factors have been implicated in
epithelial cancer progression [108,109,133,135]. Recently, Ishii et al. reported that CAFs derived from
human lung cancer specimens retain their enhanced migratory activity for some time after separation
from the cancer cells [136]. Thus, CAFs can maintain their ability to stimulate cancer progression,
suggesting that inhibition of CAF generation in tumor tissues could be a new target for controlling
primary cancer progression.
      As one candidate mediator from stromal components, secreted frizzled-related protein 1 has
been found to promote tumor–stromal interactions in PCa [137]. More recently, Ao et al. reported
that prostatic CAFs express high levels of stromal cell-derived factor-1 (also known as chemokine
(C-X-C motif) ligand 12), and the stromal cell-derived factor-1/CXCR4 pathway between cancer cells
and stromal cells could be involved in tumorigenesis of PCa [135].
J. Clin. Med. 2018, 7, 68                                                                           10 of 21

      ADT for patients with advanced PCa is intended to downregulate the concentration of circulating
androgens or to block the transcriptional activation of AR [138]. Tumor stroma surrounding cancer cells
is enriched in fibroblasts secreting AR-stimulating factors, VEGF, and TGF-β [139]. Previous studies
have indicated that a number of growth factors and cytokines, including EGF, FGF7/KGF, IGF1,
and IL-6, stimulate AR signaling and PSA expression in the context of androgen deficiency [139–142].
We have already reported that stromal remodeling after castration is accompanied by changes in the
expression levels of these growth factors in the prostate [19]. Importantly, most fibroblastic cells in
the prostate stroma are negative for AR [143,144], and the phenotypes of human PCa fibroblastic
stromal cells are strongly heterogeneous [96]. Several studies have reported that androgen-sensitive
and -insensitive interactions between cancer cells and stromal cells determine how PCa cells respond
to androgen ablation [116,132]. In a low-androgen environment, aberrant activation between cancer
cells and stromal cells may be an important mechanism controlling AR activity and AR-regulated
PSA expression.
      Soluble factors such as growth factors and cytokines derived from fibroblasts directly affect AR
stimulation and AR-regulated PSA expression in the context of androgen deprivation. Shigemura et al.
showed that conditioned medium from co-cultures of LNCaP cells and human prostate stromal
fibroblasts induced PSA promoter reporter activity, ERK phosphorylation, and AR phosphorylation in
LNCaP cells in vitro [145]. In our lab, Sasaki et al. also showed that fibroblasts directly affected PSA
expression and the activation of Stat3, but not Akt or ERK, in LNCaP cells co-cultured in vitro [146].
In this study, we confirmed that EGF, IGF1, and IL-6 stimulated the expression of AR and PSA
in LNCaP cells, suggesting that soluble factors derived from fibroblasts may function similarly to
androgen in the absence of androgen. Thus, a heterogeneous combination of growth factors and
cytokines derived from fibroblasts could be responsible for AR stimulation of PCa cells in the context
of androgen deprivation.

11. Can We Discover New Biomarkers from Heterogeneous Stroma in PCa?
       As described here, the tumor stroma is strongly heterogeneous. In our lab, Sasaki et al. formed
 a hypothesis regarding the role of fibroblasts in patients with PCa [146]. First, ‘protective’ fibroblasts
prolong the duration of the blood supply in the tumors. Second, ‘protective’ fibroblasts cause the
persistent stimulation of AR in PCa cells, preventing acute loss of AR function in PCa cells under
ADT. In contrast, tumor-promoting ‘aggressive’ fibroblasts (i.e., CAFs) have also been identified [38].
 CAFs surround cancer cells to support their survival and proliferation in a paracrine fashion.
 On the other hand, Hayashi et al. reported that rat UGM, which has features similar to CAFs
 but with the biological function of promoting the development, differentiation, and ultimately
 growth/quiescence of the prostate, elicited a reduction in the tumorigenic potential of Dunning
prostatic adenocarcinoma [147]. Recent studies have also demonstrated that normal human fibroblasts
 can inhibit the proliferation of tumor cells [148,149]. We hypothesize that these ‘protective’ fibroblasts
 can also preserve the AR dependence of PCa cells under ADT. Moreover, Banerjee et al. demonstrated
 that epigenetic changes in prostatic fibroblasts caused DNA damage, mediating prostate tumor
progression [150]. Thus, androgen deficiency may contribute to the reciprocal transfer of fibroblasts
 between the ‘protective’ and ‘aggressive’ states. Although Ayala et al. evaluated the potential of
quantifying reactive stromal elements to predict disease progression [90], we suggest that the quality
 of fibroblasts is also an important factor that can be used to distinguish between ‘aggressive’ and
‘protective’ fibroblasts, which may be determined by evaluating a combination of stromal biomarkers.
 Future studies are needed to identify the specific profile of the stroma-derived factors responsible for
 disease progression.
J. Clin. Med. 2018, 7, 68                                                                          11 of 21

12. Role of CAF-Derived Exosomal miRNAs in Aberrant Activation of Tumor–Stromal
Interactions
     Tumor–stromal interactions are a dynamic process that involves not only direct cell–cell contact
via cell surface adhesion molecules, but also indirect cell–cell communication via soluble factors.
Activated fibroblasts such as CAFs communicate with adjacent cancer cells via soluble factors secreted
into the extracellular space [151,152]. Of course, this is only one mechanism of tumor–stromal
interactions, and others may exist in parallel, such as membrane-derived exosomes. In addition to
soluble factors, membrane-derived exosomes have been reported to modulate tumor progression [134].
     In cancer, CAFs aberrantly secrete large amounts of exosomes to transport stromal
paracrine signals including miRNAs within the tumor microenvironment [134]. Exosomes are
membrane-enclosed extracellular vesicles (EVs) that contribute to tumor progression. EVs are
classified into three main types according to size and biogenesis: exosomes (30–100 nm), microvesicles
(100–1000 nm), and oncosomes (1–10 µm). The discovery of exosomes provides novel insights into
indirect cell–cell communication between cancer cells and CAFs. Exosomes play critical roles in
several aspects of tumor progression, including growth, invasion, angiogenesis, and metastasis.
Exosomes contain a wide variety of bioactive molecules including signal peptides, lipids, miRNAs,
mRNAs, and DNA [134]. Interestingly, miRNAs are stably transferred by exosomes to recipient cells
where they modulate gene expression, resulting in functional effects on cell fate.
     miRNAs are small (~20–23 nucleotides) non-coding RNAs that negatively regulate the expression
of specific target genes, including tumor suppressors and oncogenes, at the transcriptional and
translational levels [153]. In addition, target genes of miRNAs affect the fates of recipient cells
including proliferation, differentiation, adhesion, and migration, e.g., miRNAs reciprocally regulate
TGF-β signaling during tumor progression [154]. CAF-derived exosomal miRNAs affect the cell fates
of recipient cancer cells. In addition to soluble factors such as growth factors, cytokines, and ECM
proteins, exosomal miRNAs play a critical role in indirect cell–cell communication. Recently, several
studies have reported that the inhibition of miRNAs can serve as a novel therapeutic strategy for
treating or effectively managing PCa [155–158].
     In addition to the role of miRNAs as therapeutic targets, circulating exosomal miRNAs have
potential as liquid biopsy and noninvasive biomarkers for early detection and diagnosis [134].
Exosomes are also found abundantly in bodily fluids including blood, urine, cerebrospinal fluid,
breast milk, saliva, ascites fluid, and amniotic fluid [159,160]. The composition of miRNAs in circulating
exosomes is similar to that found in their originating cells [161,162]. In contrast, Schageman et al.
reported that levels of certain RNA sequences were substantially different between exosomes and the
parental serum samples [163]. These reports suggest the possibility of using disease-specific exosomal
miRNA signatures in bodily fluids as unique diagnostic markers.
     Regarding the use of miRNA expression signatures as biomarkers, several studies have recently
reported specific miRNA expression profiles in different bodily fluid samples derived from patients
with BPH or PCa (Table 1). Importantly, Cochetti et al. reported that different levels of serum miRNAs
between PCa and BPH may be reliable candidates for developing minimally invasive biomarkers as
diagnostic and prognostic tools [164]. In general, serum PSA is currently the most useful biomarker
to detect PCa, whereas an increase in serum PSA levels is also observed in BPH or inflammation
in the prostate. PSA is an androgen-regulated serine protease produced in both normal luminal
epithelial cells and well-differentiated PCa cells. ADT for patients with advanced PCa is intended to
downregulate the concentration of circulating androgens or to block AR signals, leading to a reduction
in serum PSA levels. In our previous work, Sasaki et al. found that the PSA kinetics after ADT were
not an accurate prognostic marker when we regarded serum PSA levels after ADT as the number of
viable cancer cells [165,166].
     In contrast to normal luminal epithelial cells or well-differentiated PCa cells, most fibroblasts
are AR negative. In addition, the stromal compartment of a tumor is genetically more stable than
the cancer compartment [109]. Several studies have demonstrated changes in miRNA expression
J. Clin. Med. 2018, 7, 68                                                                                       12 of 21

in the stromal compartment of PCa [92,167,168]. For example, Josson et al. demonstrated that
miR-409-3p was significantly elevated in the stroma of patients with a high (>7) versus low Gleason
score (
J. Clin. Med. 2018, 7, 68                                                                                        13 of 21

4.    Cunha, G.R. Epithelial-stromal interactions in development of the urogenital tract. Int. Rev. Cytol. 1976, 47,
      137–194. [PubMed]
5.    Ishii, K.; Imanaka-Yoshida, K.; Yoshida, T.; Sugimura, Y. Role of stromal tenascin-C in mouse prostatic
      development and epithelial cell differentiation. Dev. Biol. 2008, 324, 310–319. [CrossRef] [PubMed]
6.    Hayward, S.W.; Rosen, M.A.; Cunha, G.R. Stromal-epithelial interactions in the normal and
      neoplastic prostate. Br. J. Urol. 1997, 79 (Suppl. 2), 18–26. [CrossRef] [PubMed]
7.    Cordeiro, R.S.; Scarano, W.R.; Campos, S.G.; Santos, F.C.; Vilamaior, P.S.; Goes, R.M.; Taboga, S.R.
      Androgen receptor in the mongolian gerbil ventral prostate: Evaluation during different phases of postnatal
      development and following androgen blockage. Micron 2008, 39, 1312–1324. [CrossRef] [PubMed]
8.    Chang, S.M.; Chung, L.W. Interaction between prostatic fibroblast and epithelial cells in culture: Role
      of androgen. Endocrinology 1989, 125, 2719–2727. [CrossRef] [PubMed]
9.    Ohlson, N.; Bergh, A.; Stattin, P.; Wikstrom, P. Castration-induced epithelial cell death in human prostate
      tissue is related to locally reduced IGF-1 levels. Prostate 2007, 67, 32–40. [CrossRef] [PubMed]
10.   Takayama, K.I.; Misawa, A.; Inoue, S. Significance of microRNAs in androgen signaling and prostate
      cancer progression. Cancers 2017, 9, 102. [CrossRef] [PubMed]
11.   Hayward, S.W.; Cunha, G.R. The prostate: Development and physiology. Radiol. Clin. N. Am. 2000, 38, 1–14.
      [CrossRef]
12.   Cunha, G.R.; Ricke, W.; Thomson, A.; Marker, P.C.; Risbridger, G.; Hayward, S.W.; Wang, Y.Z.;
      Donjacour, A.A.; Kurita, T. Hormonal, cellular, and molecular regulation of normal and neoplastic
      prostatic development. J. Steroid Biochem. Mol. Biol. 2004, 92, 221–236. [CrossRef] [PubMed]
13.   Sugimura, Y.; Foster, B.A.; Hom, Y.K.; Lipschutz, J.H.; Rubin, J.S.; Finch, P.W.; Aaronson, S.A.; Hayashi, N.;
      Kawamura, J.; Cunha, G.R. Keratinocyte growth factor (KGF) can replace testosterone in the ductal branching
      morphogenesis of the rat ventral prostate. Int. J. Dev. Biol. 1996, 40, 941–951. [PubMed]
14.   Cunha, G.R.; Hayward, S.W.; Dahiya, R.; Foster, B.A. Smooth muscle-epithelial interactions in normal and
      neoplastic prostatic development. Acta Anat. (Basel) 1996, 155, 63–72. [CrossRef] [PubMed]
15.   Shibata, Y.; Ito, K.; Suzuki, K.; Nakano, K.; Fukabori, Y.; Suzuki, R.; Kawabe, Y.; Honma, S.; Yamanaka, H.
      Changes in the endocrine environment of the human prostate transition zone with aging: Simultaneous
      quantitative analysis of prostatic sex steroids and comparison with human prostatic histological composition.
      Prostate 2000, 42, 45–55. [CrossRef]
16.   English, H.F.; Santen, R.J.; Isaacs, J.T. Response of glandular versus basal rat ventral prostatic epithelial cells
      to androgen withdrawal and replacement. Prostate 1987, 11, 229–242. [CrossRef] [PubMed]
17.   Antonioli, E.; Cardoso, A.B.; Carvalho, H.F. Effects of long-term castration on the smooth muscle cell
      phenotype of the rat ventral prostate. J. Androl. 2007, 28, 777–783. [CrossRef] [PubMed]
18.   Shidaifat, F.; Daradka, M.; Al-Omari, R. Effect of androgen ablation on prostatic cell differentiation in dogs.
      Endocr. Res. 2004, 30, 327–334. [CrossRef] [PubMed]
19.   Kato, M.; Ishii, K.; Iwamoto, Y.; Sasaki, T.; Kanda, H.; Yamada, Y.; Arima, K.; Shiraishi, T.; Sugimura, Y.
      Activation of FGF2-FGFR signaling in the castrated mouse prostate stimulates the proliferation of basal
      epithelial cells. Biol. Reprod. 2013, 89, 81. [CrossRef] [PubMed]
20.   Vilamaior, P.S.; Taboga, S.R.; Carvalho, H.F. Modulation of smooth muscle cell function: Morphological
      evidence for a contractile to synthetic transition in the rat ventral prostate after castration. Cell Biol. Int.
      2005, 29, 809–816. [CrossRef] [PubMed]
21.   Sugimura, Y.; Cunha, G.R.; Donjacour, A.A. Morphological and histological study of castration-induced
      degeneration and androgen-induced regeneration in the mouse prostate. Biol. Reprod. 1986, 34, 973–983.
      [CrossRef] [PubMed]
22.   Tuohimaa, P.; Niemi, M. The effect of testosterone on cell renewal and mitotic cycles in sex accessory glands
      of castrated mice. Acta Endocrinol. (Copenh.) 1968, 58, 696–704. [CrossRef] [PubMed]
23.   Morley, A.; Wright, N.; Appleton, D.; Alison, M. A cytokinetic analysis of the proliferative response to
      androgen in the prostatic complex of the castrated mouse. Biochem. Soc. Trans. 1973, 1, 1081–1084. [CrossRef]
      [PubMed]
24.   Leong, K.G.; Wang, B.E.; Johnson, L.; Gao, W.Q. Generation of a prostate from a single adult stem cell. Nature
      2008, 456, 804–808. [CrossRef] [PubMed]
25.   Isaacs, J.T.; Coffey, D.S. Etiology and disease process of benign prostatic hyperplasia. Prostate Suppl. 1989, 2,
      33–50. [CrossRef] [PubMed]
J. Clin. Med. 2018, 7, 68                                                                                     14 of 21

26.   Isaacs, J.T. Prostate stem cells and benign prostatic hyperplasia. Prostate 2008, 68, 1025–1034. [CrossRef]
      [PubMed]
27.   Alcaraz, A.; Hammerer, P.; Tubaro, A.; Schroder, F.H.; Castro, R. Is there evidence of a relationship between
      benign prostatic hyperplasia and prostate cancer? Findings of a literature review. Eur. Urol. 2009, 55, 864–873.
      [CrossRef] [PubMed]
28.   Bostwick, D.G.; Cooner, W.H.; Denis, L.; Jones, G.W.; Scardino, P.T.; Murphy, G.P. The association of benign
      prostatic hyperplasia and cancer of the prostate. Cancer 1992, 70, 291–301. [CrossRef]
29.   Berry, S.J.; Coffey, D.S.; Walsh, P.C.; Ewing, L.L. The development of human benign prostatic hyperplasia
      with age. J. Urol. 1984, 132, 474–479. [CrossRef]
30.   Fujikawa, S.; Matsuura, H.; Kanai, M.; Fumino, M.; Ishii, K.; Arima, K.; Shiraishi, T.; Sugimura, Y.
      Natural history of human prostate gland: Morphometric and histopathological analysis of Japanese men.
      Prostate 2005, 65, 355–364. [CrossRef] [PubMed]
31.   Ishigooka, M.; Hayami, S.; Hashimoto, T.; Suzuki, Y.; Katoh, T.; Nakada, T. Relative and total volume of
      histological components in benign prostatic hyperplasia: Relationships between histological components
      and clinical findings. Prostate 1996, 29, 77–82. [CrossRef]
32.   Ichiyanagi, O.; Sasagawa, I.; Ishigooka, M.; Suzuki, Y.; Nakada, T. Morphometric analysis of symptomatic
      benign prostatic hyperplasia with and without bladder outlet obstruction. Urol. Res. 2000, 28, 29–32.
      [CrossRef] [PubMed]
33.   Djavan, B.; Marberger, M. A meta-analysis on the efficacy and tolerability of alpha1-adrenoceptor antagonists
      in patients with lower urinary tract symptoms suggestive of benign prostatic obstruction. Eur. Urol. 1999, 36,
      1–13. [CrossRef] [PubMed]
34.   Smith, P.; Rhodes, N.P.; Ke, Y.; Foster, C.S. Influence of the alpha1-adrenergic antagonist, doxazosin, on
      noradrenaline-induced modulation of cytoskeletal proteins in cultured hyperplastic prostatic stromal cells.
      Prostate 1999, 38, 216–227. [CrossRef]
35.   Justulin, L.A., Jr.; Delella, F.K.; Felisbino, S.L. Doxazosin reduces cell proliferation and increases collagen
      fibers in rat prostatic lobes. Cell Tissue Res. 2008, 332, 171–183. [CrossRef] [PubMed]
36.   Imamura, T.; Ishii, K.; Kanda, H.; Arase, S.; Yoshio, Y.; Hori, Y.; Soga, N.; Kise, H.; Arima, K.; Sugimura, Y.
      Structural changes in alpha1-adrenoceptor antagonist-treated human prostatic stroma. Clin. Exp. Med.
      2010, 10, 99–106. [CrossRef] [PubMed]
37.   Verrecchia, F.; Mauviel, A. Transforming growth factor-beta and fibrosis. World J. Gastroenterol. 2007, 13,
      3056–3062. [CrossRef] [PubMed]
38.   Olumi, A.F.; Grossfeld, G.D.; Hayward, S.W.; Carroll, P.R.; Tlsty, T.D.; Cunha, G.R. Carcinoma-associated
      fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999, 59, 5002–5011.
      [PubMed]
39.   Bhowmick, N.A.; Chytil, A.; Plieth, D.; Gorska, A.E.; Dumont, N.; Shappell, S.; Washington, M.K.;
      Neilson, E.G.; Moses, H.L. TGF-beta signaling in fibroblasts modulates the oncogenic potential of
      adjacent epithelia. Science 2004, 303, 848–851. [CrossRef] [PubMed]
40.   Bierhoff, E.; Vogel, J.; Benz, M.; Giefer, T.; Wernert, N.; Pfeifer, U. Stromal nodules in benign
      prostatic hyperplasia. Eur. Urol. 1996, 29, 345–354. [CrossRef] [PubMed]
41.   Pradhan, B.K.; Chandra, K. Morphogenesis of nodular hyperplasia–prostate. J. Urol. 1975, 113, 210–213.
      [CrossRef]
42.   McNeal, J.E. Origin and evolution of benign prostatic enlargement. Investig. Urol. 1978, 15, 340–345.
43.   Norman, J.T.; Cunha, G.R.; Sugimura, Y. The induction of new ductal growth in adult prostatic epithelium in
      response to an embryonic prostatic inductor. Prostate 1986, 8, 209–220. [CrossRef] [PubMed]
44.   De Marzo, A.M.; Nelson, W.G.; Meeker, A.K.; Coffey, D.S. Stem cell features of benign and malignant prostate
      epithelial cells. J. Urol. 1998, 160, 2381–2392. [CrossRef]
45.   Collins, A.T.; Habib, F.K.; Maitland, N.J.; Neal, D.E. Identification and isolation of human prostate epithelial
      stem cells based on alpha(2)beta(1)-integrin expression. J. Cell Sci. 2001, 114, 3865–3872. [PubMed]
46.   Wang, Y.; Hayward, S.; Cao, M.; Thayer, K.; Cunha, G. Cell differentiation lineage in the prostate.
      Differentiation 2001, 68, 270–279. [CrossRef] [PubMed]
47.   Hudson, D.L. Epithelial stem cells in human prostate growth and disease. Prostate Cancer Prostatic Dis.
      2004, 7, 188–194. [CrossRef] [PubMed]
J. Clin. Med. 2018, 7, 68                                                                                       15 of 21

48.   Kurita, T.; Medina, R.T.; Mills, A.A.; Cunha, G.R. Role of p63 and basal cells in the prostate. Development
      2004, 131, 4955–4964. [CrossRef] [PubMed]
49.   Maitland, N.J.; Frame, F.M.; Polson, E.S.; Lewis, J.L.; Collins, A.T. Prostate cancer stem cells: Do they have
      a basal or luminal phenotype? Horm. Cancer 2011, 2, 47–61. [CrossRef] [PubMed]
50.   Cleary, K.R.; Choi, H.Y.; Ayala, A.G. Basal cell hyperplasia of the prostate. Am. J. Clin. Pathol. 1983, 80,
      850–854. [CrossRef] [PubMed]
51.   Dermer, G.B. Basal cell proliferation in benign prostatic hyperplasia. Cancer 1978, 41, 1857–1862. [CrossRef]
52.   Rioux-Leclercq, N.C.; Epstein, J.I. Unusual morphologic patterns of basal cell hyperplasia of the prostate.
      Am. J. Surg. Pathol. 2002, 26, 237–243. [CrossRef] [PubMed]
53.   Wang, S.; Garcia, A.J.; Wu, M.; Lawson, D.A.; Witte, O.N.; Wu, H. Pten deletion leads to the expansion of
      a prostatic stem/progenitor cell subpopulation and tumor initiation. Proc. Natl. Acad. Sci. USA 2006, 103,
      1480–1485. [CrossRef] [PubMed]
54.   Mirosevich, J.; Bentel, J.M.; Zeps, N.; Redmond, S.L.; D’Antuono, M.F.; Dawkins, H.J. Androgen receptor
      expression of proliferating basal and luminal cells in adult murine ventral prostate. J. Endocrinol. 1999, 162,
      341–350. [CrossRef] [PubMed]
55.   Sugimura, Y.; Sakurai, M.; Hayashi, N.; Yamashita, A.; Kawamura, J. Age-related changes of the prostate
      gland in the senescence-accelerated mouse. Prostate 1994, 24, 24–32. [CrossRef] [PubMed]
56.   Leav, I.; Schelling, K.H.; Adams, J.Y.; Merk, F.B.; Alroy, J. Role of canine basal cells in prostatic post natal
      development, induction of hyperplasia, sex hormone-stimulated growth; and the ductal origin of carcinoma.
      Prostate 2001, 47, 149–163. [CrossRef] [PubMed]
57.   Waltregny, D.; Leav, I.; Signoretti, S.; Soung, P.; Lin, D.; Merk, F.; Adams, J.Y.; Bhattacharya, N.; Cirenei, N.;
      Loda, M. Androgen-driven prostate epithelial cell proliferation and differentiation in vivo involve the
      regulation of p27. Mol. Endocrinol. 2001, 15, 765–782. [CrossRef] [PubMed]
58.   Nishi, N.; Oya, H.; Matsumoto, K.; Nakamura, T.; Miyanaka, H.; Wada, F. Changes in gene expression of
      growth factors and their receptors during castration-induced involution and androgen-induced regrowth of
      rat prostates. Prostate 1996, 28, 139–152. [CrossRef]
59.   Kyprianou, N.; Isaacs, J.T. Expression of transforming growth factor-beta in the rat ventral prostate during
      castration-induced programmed cell death. Mol. Endocrinol. 1989, 3, 1515–1522. [CrossRef] [PubMed]
60.   Wang, G.M.; Kovalenko, B.; Huang, Y.; Moscatelli, D. Vascular endothelial growth factor and angiopoietin
      are required for prostate regeneration. Prostate 2007, 67, 485–499. [CrossRef] [PubMed]
61.   Donjacour, A.A.; Thomson, A.A.; Cunha, G.R. FGF-10 plays an essential role in the growth of the fetal prostate.
      Dev. Biol. 2003, 261, 39–54. [CrossRef]
62.   Lin, Y.; Wang, F. Fgf signalling in prostate development, tissue homoeostasis and tumorigenesis. Biosci. Rep.
      2010, 30, 285–291. [CrossRef] [PubMed]
63.   Risbridger, G.; Wang, H.; Young, P.; Kurita, T.; Wang, Y.Z.; Lubahn, D.; Gustafsson, J.A.; Cunha, G. Evidence
      that epithelial and mesenchymal estrogen receptor-alpha mediates effects of estrogen on prostatic epithelium.
      Dev. Biol. 2001, 229, 432–442. [CrossRef] [PubMed]
64.   Tunn, U.; Senge, T.; Schenck, B.; Neumann, F. Biochemical and histological studies on prostates in castrated
      dogs after treatment with androstanediol, oestradiol and cyproterone acetate. Acta Endocrinol. (Copenh.)
      1979, 91, 373–384. [CrossRef] [PubMed]
65.   Timms, B.G.; Howdeshell, K.L.; Barton, L.; Bradley, S.; Richter, C.A.; vom Saal, F.S. Estrogenic chemicals
      in plastic and oral contraceptives disrupt development of the fetal mouse prostate and urethra. Proc. Natl.
      Acad. Sci. USA 2005, 102, 7014–7019. [CrossRef] [PubMed]
66.   Ogura, Y.; Ishii, K.; Kanda, H.; Kanai, M.; Arima, K.; Wang, Y.; Sugimura, Y. Bisphenol a induces permanent
      squamous change in mouse prostatic epithelium. Differentiation 2007, 75, 745–756. [CrossRef] [PubMed]
67.   Arase, S.; Ishii, K.; Igarashi, K.; Aisaki, K.; Yoshio, Y.; Matsushima, A.; Shimohigashi, Y.; Arima, K.;
      Kanno, J.; Sugimura, Y. Endocrine disrupter bisphenol a increases in situ estrogen production in the mouse
      urogenital sinus. Biol. Reprod. 2011, 84, 734–742. [CrossRef] [PubMed]
68.   Ciana, P.; Ghisletti, S.; Mussi, P.; Eberini, I.; Vegeto, E.; Maggi, A. Estrogen receptor alpha, a molecular
      switch converting transforming growth factor-alpha-mediated proliferation into differentiation in
      neuroblastoma cells. J. Biol. Chem. 2003, 278, 31737–31744. [CrossRef] [PubMed]
69.   Kaplan, P.J.; Leav, I.; Greenwood, J.; Kwan, P.W.; Ho, S.M. Involvement of transforming growth factor alpha
      (TGFalpha) and epidermal growth factor receptor (EGFR) in sex hormone-induced prostatic dysplasia and
You can also read